Drugs, Neurochemistry and Transmission of Nervous Systems Flashcards

1
Q

What are the fundamental principles of chemical neurotransmission at a synapse?

A
  1. Synthesis of NT
  2. Storage of NT
  3. Release of NT
  4. Reuptake of NT
  5. Metabolism of NT
  6. Receptor Interaction
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

What percentage of NA in synaptic clefts is reuptaken and recycled?

A

Approximatley 90-95% of synaptic NA is recycled - all most of it neuronally reuptaken and a small portion taken up extraneuronally (e.g. by glial cells)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

NA regulatory feedback is achieved pre-synaptically via what type of receptor?

A

a2 adrenoceptors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

What are monamine oxidases and catechol-o-methyltransferases (COMT)?

A

Monoamine Oxidases (MAO) are enzymes responsible for the breakdown of monoamines such as NA, Adr and dopamine. They are involved in the synaptic metabolism of recycled NA - mainly presynaptically but some extraneurally.

Catechol-o-methyltransferases (COMT) are enzymes that breakdown catecholamine such as dopamine, adrenaline and noradrenaline. They are involved in the synaptic metabolism of recycled NA - mainly extraneurally.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Describe the pathway of catecholamine biosynthesis

A

The only difference between cells capable of producing adrenaline from noradrenaline or dopamine is the presence of the later enzymes withint he pathway.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What are the dose dependent effects of cocaine?

A
  1. Intense euphoria + craving for more drug
  2. Psychological and physical dependence
    * psychotic symptoms, depression, anxiety and fatigue
  3. Cardiovascular effect
    * hypertension, tachycardia, coronary vasospasm, dysrhythmia, convulsions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

How do amphetamines and ephadrines affect NA storage in neurons?

A

Amphetamines and ephadrines are indirectly acting sympathomimetics that act on the presynaptic storage of NA.

These drugs displace NA from its storage vesicles. This results in the non-exocytotic release of NA into the synapse via catecholamine channels.

This causes increased activation of a- or b-adrenoceptors both peripherally and centrally.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

What functions are noradrenergic and dopaminergic pathways associated with in the CNS?

A

Noradrenergic pathways are involved in stimulant, mood, appetite and cardiovascular regulation

Dopamine is considered nothing more than a precursor or NA in the periphery, but in the CNS dopaminergic pathways are involved in:

  • Movement - Parkinson’s results from depletion of dopamine in basal ganglia
  • Behaviour - Schizophrenia results from changes in dopamin rich areas
  • Dependence - Dopaminergic neurons innervate known dependence nuclei in nucleus accumbens and ventral tegmental area
  • Pituitary Function - Dopamin results in prolactin secretion
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Discuss the selectivity of cocaine

A

Cocaine blocks the reuptake of noradrenaline, dopamine and serotonin at synapses in the CNS

The dopaminergic actions are linked to dependence.

Noradrenergic and serotonergic actions are leads for anti-depressant drugs in development.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What is the first line drug treatment of Parkinson’s Disease?

A

Parkinson’s Disease

Is characterised by the degeneration of dopaminergic pathways

Treatment with:

  • L-DOPA + peripheral DOPA Decarboxylase Inhibitor
    • Increases the biosynthesis pathway of dopamine in the CNS; while mitigating peripheral side effects of dopamine in the periphery.
  • Secondary: MAO B inhibitors, dopamine receptor agonists and muscarinci receptor antagonists
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What is the first line drug treatment of Huntington’s Disease?

A

Huntington’s Disease

Is characterised by a deficiency in GABA within the CNS

Treatment:

  • GABA agonist Baclofen
  • Dopamine antagonists Chloropromazine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

What are the three divisions of the autonomic nervous system?

A
  1. Sympathetic NS
  2. Parasympathetic NS
  3. Enteric NS
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Do autonomic pathways originate in cervical or lower lumbar regions of the spinal cord?

A

No; there are no autonomic pathways arising from the cervical or lower lumbar enlargements

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

From which regions of the spinal cord do sympathetic and parasympathetic outputs arise respectively?

A

Sympathetic NS

Thoraco-lumbar spinal segments (but not lower lumbar)

Parasympathetic NS

Craniosacral spinal segments

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Compare and contrast the position of sympathetic and parasympathetic ganglia

A

Sympathetic ganglia are typically located more distant to their organs of innervation. Most pre-ganglionic nerves of the sympathetic NS synapse to the paravertebral ganglia (sympathetic chain) that is parallel to the spinal column. These ganglia are the primary source of vasoconstrictor neurons.

Some preganglionic neurons, like pelvic visceral neurons, go through the paravertebral ganglia but do not synapse - instead synapsing on post-ganglionic neurons at **prevertebral ganglia **closer to organ of innervation.These ganglia are the primary source of non-vascular smooth muscle innervation.

Parasympathetic ganglia are generally located closer to or within individual organs.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Which level of neurons in the sympathetic, parasympathetic and somatic NS’s are myelinated?

A

All preganglionic nerves of the autonomic NS are lightly myelinated or unmyelinated.

Postganglionic nerves of the autonomic NS are all unmyelinated.

Somatic motor nerves are variable in myelination

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

Which neurotransmitters are considered ‘classical’ transmitters of the autonomic nervous system?

Provide examples of common ‘non-classical’ transmitters

A

Classical transmitters applies to the dominant neurotransmitters ACh and NA.

Non classical transmitters include:

  • ATP
  • Nitric Oxide
  • Neuropeptides - Substance P, Neuropeptide Y

Often there is more than one transmitter being released at any one time = co-transmission.

Glutamate and GABA are not significant transmitters in the autonomic nervous system.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

What is special about autonomic neurotransmission in the organs?

A
  1. No easily identifiable synaptic junctions
  2. More than 1 NT release site per axon
  3. Receptors can be expressed remotely from the synapse via extrajunctional receptors
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Where in the spinal cord are preganglionic neurons located within the spinal cord?

A

Cholinergic preganglionic neurons of the sympathetic nervous system are located in the intermediolateral cell column of spinal cord grey matter.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

With reference to the concepts of divergence and convergence, discuss the differences in post-ganglionic neurons in the sympathetic NS

A

Prevertebral ganglion neurons integrate more signals from multiple inputs (particularly the enteric nervous system) and have mor complex dendritic organise to allow greater convergence of information.

Up to 200 paravertebral ganglionic neurons can be activated by one preganglionic neuron - illustrating divergence to produce larger downstream effects.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

What are the common functions of sympathetic nervous system activation?

A

Sympathetic activation is characterised by the ‘flight-or-flight’ set of responses; including:

  • Increased heart rate
  • Increased contractility of the heart
  • Increased blood flow to skeletal muscle
  • Decreased blood flow to gut
  • Decreased gut motility
  • Relaxation of airways

Additionally, the preganglionic sympathetics can activate the adrenal gland to release ACh and Adrenaline into the circulation and produce broad systemic activation of adrenoceptors throughout the body

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

Discuss some examples of preganglionic neurons of the parasympathetic NS that arise from the cranial compartment of the nervous system.

A

Edinger-Westfal Nucleus

Projects to the ciliary ganglion to control sphincter pupillae and ciliary muscle.

Salivatory Nuclei

To submandibular, sphenopalatine and optic ganglia to control lacrimal, salivary, sublingual, nasal and palatine glands

Dorsal Motor Nucleus of Vagus and Nucleus Ambiguss

To microganglia near and on outer surface of thoracic and abdominal organs for visceral functions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What is special about autonomic supply to the pelvic organs?

A

Autonomic supply to pelvic organs is via splanchnic nerves to pelvic ganglia that comprise the **pelvic plexus **overlying or within pelvic organs.

These parasympathetic ganglia have abnormally long axons - leaving them vulnerable to surgically induced injury.

These pelvic ganglia are mixed ganglia - also containing many sympathetic neurons.

Pelvic ganglia are mere relay stations - little integration/few dendrites present

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

Where in the spinal cord would you find cell bodies of preganglionic parasympathetic neurons?

A

Preganglionic parasympathetic neurons are located laterally within the intermediate grey zone of the sacral spinal cord

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

What are the common features of parasympathetic activation?

A

Parasympathetic activation invokes a ‘rest and digest’ set of functions; including:

  • Decreased heart rate
  • Decreased contractility of the heart
  • Increased gut motility
  • Constriction of airways

Compared with sympathetic pathways, parasympathetic have:

  • lower ratios of pre:post ganglionic neurons (less divergence)
  • Parasympathetic ganglia are simple relay stations that don’t integrate or coordinate information
  • No equivalent of adrenal gland for systemic activation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

How can you definitively distinguish between sympathetic and parasympathetic nerves?

A

The only unequivocal definition between sympathetic and parasympathetic nerves is the location of preganglionic fibres

Sympathetic: thoracic + lumbar spinal cord

Parasympathetic: cranial nuclei + sacral spinal cord

Other properties are unreliable and too many exceptions exist to be used as general rules of distinction; e.g.:

  • Neurotransmitters
  • Strucutre of ganglions neurons
  • Physiological effects
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

Describe a typical autonomic reflex

A

Visceral afferent (sensory) neurons provide input to local interneurons and projection neurons (either ascending or efferent) within the spinal and brainstem circuits.

Most autonomic reflexes involve the brain (supraspinal reflex); rather than only the spinal cord(spinal reflex)

Lesions that disrupt ascending and descending connections with the brain can cause disruption to reflexes involving: bladder, sexual function, cardiovascular and thermal regulation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

How is the hypothalamus involved in the coordination of autonomic output?

A

The hypothalamus initiates and coordinates an appropriate autonomic responses involving:

  • Fight or flight
  • Feeding
  • Thermoregulation
  • Circadian rhythms
  • Water balance
  • Sexual function

Hypothalamus compares current situations with biological set points; adjust behavoiur and autonomic function to achieve homeostasis.

It integrates with higher cortical and limbic systems to produce appropriate responses to emotion, fear, anxiety and motivations.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

What is the role of the nucleus of the solitary tract in autonomic NS regulation?

A

The caudal part of the nucleus of the solitary tract (NTS) is located in the medulla.

It is a major integrative centre for autonomic function

It recieves input from visceral afferents and distributes information to either:

  1. local reflexes that control organ/tissue function via preganglionic neurons; or
  2. higher cortical and subcortical centres for more complex responses (e.g hypothalamus, thalamus and cortex)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

What are local anaesthetics?

and

What classes of local anaesthetics exist?

A

Local anaesthetics are drugs that reversibly block the conduction of nerve impulses at the axonal membrane via Na+ channel inhibition

Local anaesthetics tend to be weak bases with different onset, duration and toxicity profiles

There are three main classes of local anaesthetics:

  1. Aminoesters
  • Hydrophilic mechanism of action
  • Shorter acting (30-45 min)
  • Removed by hydrolysis via esterases
  • e.g. procaine
  1. Aminoamides
  • Hydrophilic mechanism of action
  • Longer acting (hours)
  • Removed via hepatic metabolism
  • e.g. Lignocaine and **Ropivicaine **(most common in Aus)
  1. Benzocaine
    * Hydrophobic mechanism of action
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

Where is the binding site of local anaesthetics?

A

Local anaesthetics bind S6 in transmembrane domain IV of Na+ channels

  • This binding site is intracellular - affects the mechanism of action of local anaesthetics able to enter the cell
    • Hydrophobic (benzocaine) vs Hydrophilic (aminoester and aminoamines)

Toxins (tetrodotoxin/puffer fish) bind extracellular domains of Na+ channels to cause anaesthetic action - different mode

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

Discuss the differences in sensitivity to local anaesthetics between different types of nerves.

A

Smaller nerve fibres are more sensitive to local anaesthetic effects. Thus sensitivity in:

sensory > autonomic > motor

The preferential effects on sensory nerves allows sensation to be inhibited without compromising the motor effects of the nervous system - e.g respiratory and cardiac innervation required for survival.

In sufficient concentrations, however, widespread motor impairment is likely.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

Compare the two different mechanisms of local anaesthetic action

A

There are two mechanisms of local anaesthetic actions:

Hydrophobic Pathway (non use-dependent)

The base form of the drug (B) is the predominant form (uncharged) and freely diffuses through the lipid membrane of axons as a hydrophobic molecule. It directly binds to the S6 transmembrane domain and blocks electrical conduction.

This pathway is fast acting and non-use-dependent due to the rapid ease of binding. Benzocaine is an example

Hydrophilic Pathway (use dependent)

The predominant drug form is ionised (BH+) and has difficulty in diffusing through axon membrane. Small proportion is in base form (B) and this is able to diffuse through into the intracellular space. In the intracellular space base form (B) becomes charged again (BH+) and enters the channel to bind S6 - this requires the channels to be open and being ‘used’.

Thus, they are slow acting and use dependent. Examples include aminoesters and aminoamines.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

What are the general properties of local aesthetics?

A
  1. Prevent the propagation of nerve action potentials
  2. Small fibres are more sensitive (sensory > ANS > motor)
  3. Axon membrane (RMP) remains stable/unchanged
  4. Effects more pronounced in basic mediums
  • uncharged species are more active
  • at physiological p.H 7.4 more ionised forms are present
    1. Greater anaesthetic affects if the nerve is firing / being used
35
Q

Discuss local anaesthetic toxicity

A

Local anaesthetics are generally safe drugs when used by trained physicians.

They selectively bind Na+ channels and display reversible binding without nerve damage…but will affect all excitable tissues with Na+ channels.

proportional [Blood] toxicity

Cardiovascular

  • direct myocardial depression
  • depression of vasomotor centre
  • hypotension (except cocaine)

CNS

  • excitation (inhibitory fibres first affected)
  • tremor
  • convulsion
  • respiratory arrest

non-proportional [Blood] toxicity

Hypersensitivity/Allergic reactions

  • both to the anaesthetic itself and their stabilising agents
36
Q

Discuss the usefulness of topical local anaesthetics

A

There are two main forms of topical local anaesthetics:

Lozenges:

Generally used for throat drops - contain benzocaine (fast acting, readily diffusable)

Gels:

Tend to contain small concentrations of lignocaine. Generally poor absorption across skin (barrier to access) and acidic inflammatory environment inconducive to mechanism of action (ionises the drug - can’t diffuse into axon membrane)

37
Q

What are the four stages of general anaesthesia?

A

Stage 1

  • Amnesia
  • Euphoria

Stage 2

  • Excitement
  • Delirium
  • Resistance to handling

Stage 3

  • **Commence surgery **
  • Unconsciousness
  • Regular respiration
  • Decreasing eye movement

Stage 4

  • Respiratory arrest
  • Cardiac depression + arrest
38
Q

Provide examples of general anaesthetic agents

A

Inhalational:

  • Desflurane
  • Sevoflurane
  • Isoflurane

Intravenous

  • Propofol
  • Thiopentone
39
Q

What are the side effects of general anaesthetics?

A

Respiratory

  • Impaired ventilation
  • Depression of respiratory centers
  • Obstruction of airways
    • Retention of secretions in airways

Cardiovascular

  • Decreased vasomotor centre function
  • Depress contractility
  • Peripheral vasodilation
  • Cardiac arrhythmias
  • Inadequate responses to BP or CO changes
40
Q

How do general anaesthetics work?

A

General anaesthetics depress cortical processing of pain/sensory signals by inducing a loss of consciousness.

It is not known how general anaesthetics work exactly

Two leading hypothesises:

_Lipid Theory _

  • Anaesthesia is caused by volume expansion of membrane lipids (which can be reversed by pressure) that disrupts electrical signalling
  • There is an intimate correlation between anaesthetic potency and lipid solubility

Receptor Interactions

  • Anaesthetics inhibit a range of excitory receptors (glutamate and NMDA) and enhance effects on inhibitory receptors (GABA + glycine)
41
Q

How can you selectively target receptors and ion channels of the CNS to treat symptoms of epilepsy?

A

In epilepsy, there is excessive discharge of motor neurons as a result of abnormal inputs from excitatory and inhibitory interneurons

Can selectively target use-dependent pathways of the CNS

Reduce excitatory input:

  1. Limit excitatory interneuron activation
    • **Phenytonin **inhibits Na+channels of these neurons
  2. Inhibit T-type Ca2+ channels (prevents NT release) from interneurons
  3. Inhibit excitatory receptors on motor neurons

Enhance inhibitory input:

  1. Enhance GABA receptor activity
    * Benzodiazapines
42
Q

What is nociceptive pain?

A

Nociceptive pain is an adaptive, high threshold pain that acts as an early warning system against further damage.

Nociceptors are high threshold receptors of noxious stimuli - the stimulus must be of high intensity to elicit an action potential response and transmit to the CNS.

Nociceptive pain leads to the perception of pain, autonomic response to pain and withdrawl reflexes.

43
Q

What neuron fibre types are responsible for conducting nociceptive pain?

A

Nociceptive pain is conducted via two types of free nerve ending sensory afferents:

_C-fibres: _

  • fine diameter (<1.5 micrometers)
  • unmyelinated
  • slow transmission (<3m/s)
  • innervate superficial regions of dorsal horn laminae - regions 1 & 2

_Ad fibres: _

  • thick diameter (1.5-4 micrometers)
  • myelinated fibres
  • fast transmission (3-30m/s)
  • innervate region 1 & 5 of dorsal horn laminae
44
Q

By what pathway does nociceptive information transmit to the brain/within the CNS?

A

Primary C-fibre and Ad sensory afferents enter the dorsal horn and synapse onto secondary sensory afferents.

These secondary afferents decussate to the contralateral side of the spinal cord and run in the anterolateral tract to the brain.

45
Q

Can nociceptive primary afferents be stimulated without the experience of pain?

A

There are local spinal cord interneurons responsible for producing a reflex response to nociceptive afferent fibre activation - meaning there is no brain processing and no pain felt.

Thus, nociceptive fibres can be activated without feeling pain.

46
Q

What is inflammatory pain?

A

Inflammatory pain is an adaptive, low threshold pain that promotes injury repair by inducing avoidance behaviour in not using body parts due to pain hypersensitivity/tenderness.

Inflammatory structures such as macrophages, mast cells, neutrophils, granulocytes and damaged tissue itself release a range of mediators that target and sensitise nociceptor ion channels and receptors. This is **peripheral sensitisation **and leads to primary (peripheral) **hyperalgesia **

47
Q

What is the significance of TRPV1?

A

TRPV1 is a nociceptive transducer involved in the sensation of heat, capsaicin (chillis), acid and chemicals

48
Q

Contrast and compare the concepts of ‘hyperalgesia’ and ‘allodynia’

A

Hyperalgesia is an increased response to a normally painful stimulus

Allodynia is a painful response to a normally innocuous stimulus

49
Q

What is the difference between primary and secondary forms of hyperalgesia and allodynnia?

A
50
Q

Does inferred pain result from primary or secondary hyperalgesia?

A

Sensitisation of the CNS alters how primary nociceptive afferents are processed in the spinal cord.

In the presence of a sustained noxius stimulant in the periphery, secondary hyperalgesia throughout levels of the spinal cord can infer pain from a wider area than where the noxious stimulus actually is

51
Q

What is maladaptive pain?

A

Maladaptive pain is maladaptive, low threshold pain that results from disease of the nervous system. It involves marked neuroimmune responses leading to _peripheral and/or central sensitisation. _

It incorporates neuropathic pain and dysfunctional pain

Neuropathic pain results from lesions to the somatosensory system in the periphery and/or CNS

**Dysfunctional pain **occurs for no known reason e.g. migraines or neuromyalgia.

Pain is both spontaneous and stimulus dependent

52
Q

What is pain?

How does pain relate to behaviour?

A

Pain is an unpleasant sensory and emotional experience associated with actual or potential tissue damage - or described in terms of such damage

Pain is expressed in behaviour. Our behaviour os how we express and communicate pain to others (e.g. crying + avoidance); thus, pain has an important effect on behaviour

53
Q

Can pain be modulated in any way?

A

There is a descending pain modulation pathway that can block/interfere with the transmission of pain related information within the spinal cord.

The pathway runs through the anterior cingulate, prefrontal cortex, insula, **periaqueductal gray region **of the brainstem, **RVM **and down the spinal cord onto primary and secondary nociceptive afferents within the spinal cord.

This pathway is engaged by inflammation, analgesic drugs and psychophysiological states.

  • Placebo and nocebo effects can result from this pathway
54
Q

In the pshychobiological model of pain; what are the main domains of experiencing pain?

A
  1. Nociception
  2. Injury - peripheral and central sensitisation
  3. Chemical and structural - atrophy and opiodergic/dopaminergic dysfunction
  4. Context - beliefs, expectations, placebo/nocebo
  5. Mood - depression, anxiety, fear, catastophising
  6. Cognition - attention, control, re-appraisal
55
Q

What are b**enzodiazepines **indicated for?

A

Benzodiazepines are indicated for treatment:

  • Epilepsy
  • Anxiety
  • Sleep disorders
  • Premedication
    • Sedation for medical procedures
  • Acute alcohol withdrawl
56
Q

What neurotransmitters are implicated as sedative-hypnotic and anxiolytic agents?

A

Sedation-hypnotic and anxiety

GABA

  • main inhibitory transmitter in the CNS
  • GABA A ligand-gated receptor primary target

Serotonin

  • CNS prejunctional receptors

Anxiety

Noradrenaline

Neuropeptide Y

  • target of future therapies

Sedation

Histamine

  • H1 receptor antagonists are sedative
57
Q

What is anxiety?

A

Anxiety is the manifestation of “fear responses” in an anticipatory manner, independent of external events

It is associated with:

  • marked sweating
  • tachycardia
  • chest pains
  • trembling
  • choking

Beta-adrenoceptor antagonists provide some symptomatic relief to periphery symptoms but have little effect on the CNS symptoms.

Need sedative-hypnotic and anxiolytic agents.

58
Q

What are the four main clinically recognised anxiety disorders?

A
  1. Generalised anxiety states
    * excessive anxiety lacking any clear reason or focus
  2. Panic disorder
    * overwhelming fear in association with somatic signs
  3. Phobias
    * Strong fears of specific things/situations
  4. Post-traumatic stress disorder
59
Q

Profile barbituate pharmacology

A

Barbituates are general depressants that produce all levels of CNS depression -> mild sedation, surgical anaesthesia, coma and death.

It is a directly gating or channel opening agent of GABA-A receptors: it prolongs the opening of these Cl-_ channels in response to GABA_.

Increases both the sensitivity to GABA and maximum response of the GABA-A receptor

It is an effective compound; but is:

  • exceedingly toxic
    • low therapeutic index
    • inducts liver enzymes
    • abrupt withdrawl causes death
  • highly addictive

Is no longer prescribed for anxiolytics/hypnotics due to danger of overdosing, but continues to be used in controlled situations as an anaesthetic and anti-convulsant.

60
Q

Profile the pharmacology of bemzodiazepines

A

Benzodiazepines are general depressants.

Compared with barbituates, benzodiazepines demonstrate:

  • less depression of respiratory and cardiovascular centres
  • less dependence
  • safety in overdose due to the way they affect the GABA-A receptors

Benzodiazepines are allosteric modulators of the GABA-A receptor. They facilitate the opening of Cl- channels -> increasing the frequency of channel opening, but not changing the mean open time/conductance of Cl-.

Benzodiazepines increase the sensitivity of the receptor with no change in maximum response.

Disadvantages of benzodiazepines:

  • Interactions with alcohol, anti-histamines and barbituates
  • Long lasting hang-over effects (drowsiness etc)
  • Withdrawls symptoms common
  • Dependence (but less than barbituates)
    • nausea, tremor,anxiety, depression etc.
  • Tolerance development
    • need gradual dose escalation
61
Q

What are the three different pathways of allosteric modulation

A
  1. Modulation of orthosteric ligand activity
  2. Modulation of orthosteric ligand efficacy
  3. Modulation of receptor activation level
62
Q

What are the advantages of allosteric modulators?

A
  1. Ceiling of effect of inhibitors
    * increases therapeutic window
  2. Positive modulation of endogenous agonist effect rather than continuous effect of exogenous agonist
    * physiological regulation continues
  3. Great receptor subtype selectivity possible
63
Q

What are the symptomatic relief effects of benzodiazepines?

What adverse effects can occur?

A

Benzodiazepines elicit:

  • Sedation and induction of sleep
    • reduce time to fall asleep
    • increase duration of sleep
  • Reduction of anxiety and aggression
  • Reduction of muscle tone
    • anti-convulsant (but reduces coordination)
  • Obliterate memory
    • used as premedicant to surgery etc.

Side effects of benzodiazepines:

  • Drowsiness
  • Confusion
  • Impaired consciousness

These effects contribute to the ‘hang-over’ experienced after taking benzodiazepines

64
Q

What factors are considered when determining the type (short acting vs long acting) of benzodiazepine?

clue: use as a hypnotic or anxiolytic?

A
65
Q

Compare and contrast the effects of barbituates and benzodiazepines on GABA potency and efficacy

A

Both compounds potentiate the effect of GABA to GABA-A receptors.

Benzodiazepines (increased potency)

  • increase the frequency of the chloride ion channel opening at the GABA-A receptor
  • Pharmacodynamics: _This increases the potency of GABA _
    • less GABA dosage required because the GABA present has elevated affinity for the GABA-A receptor

Barbituates (increased efficacy)

  • Barbiturates produce their pharmacological effects by increasing the duration of chloride ion channel opening at the GABA-A receptor
  • Pharmacodynamics: This increases the efficacy of GABA
66
Q

Why are benzodiazepines safer when overdosing compared to barbituates?

A

The direct gating or opening of the chloride ion channel is the reason for the increased toxicity of barbiturates compared to benzodiazepines in
overdose.

Benzodiazepines increase the frequency of the chloride ion channel opening at the GABA-A receptor. As an allosteric modulator of the receptor, there is a functional ceiling of effect that benzodiaepines reach -> unable to produce more effect/harm above this level.

67
Q

Discuss some non-benzodiazepine hypnotics

A

Zolpidem

  • short term treatment of insomnia
  • short action of duration (t1/2= 4 hrs)
  • binds same benzodiazepine site

Zopiclone

  • short term treatment of insomnia
  • profile similar to benzodiazepine although structurally unrelated
  • Binds at a seperate site to BDZ at the GABA-A receptor
68
Q

Discuss a non-benzodiazepine anxiolytic

A

Buspirone

  • partial agonist at 5HT-1Areceptors
    • these are inhibitory autoreceptors regulating non-serotonergic nerve transmitter release
  • slow onset (2 weeks)
  • little dependence
  • side effects
    • nausea, diziness, headache
69
Q

What is the difference between drug dependence and drug abuse?

A

Drug Dependence

State where drug use becomes compulsive taking precedence over other needs arises because of

Drug Abuse

Use of illicit substances (or illicit use of legal substances) characterised by recurrent and clinically significant adverse consequenses.

70
Q

What circuits are involved in drug abuse and addiction?

A

Reward / Salience

  • nucleus accumbens
  • ventral pallidium

Inhibitory control

  • prefrontal cortex
  • anterior cingulate gyrus

Memory and learning

  • hippocampus
  • amygdala

Motivation / drive

  • orbitofrontal cortex
  • subcallosal cortex
71
Q

How do drugs of dependence cause dependence?

A

They increase dopamine in the nucleus accumbens

There are several key transmitters that modulate dopaminergic transmission:

  • Acetylcholine, Serotonin (5-HT), Noradrenaline
  • GABA, Glutamate
  • Opioids
72
Q

Why do people take drugs of dependence?

A

To feel good

Experience novel feelings, sensations, experiences

To feel better

Lessen anxieties, worries, fears, depression, hopelesness

Note: the same area of the brain involved in dependence are deficient in transmission in states of depression

73
Q

Discuss illicit **amphetamines **as a CNS stimulant

A

Amphetamines are indirectly acting sympathomimetics that displace DA, 5-HT or NA from vesicles in the pre-synaptic cell and elevate synaptic levels of these NT via non-exocytotic monoamine channels

**Dependence relates to increasing DA levels in the nucleus accumbens **

Effects as a CNS stimulant:

Vary with mood, personality, environment and dose

  • mood elevation, euphoria
  • increase locomotor activity
  • stereotypic behaviour to individual

Improved physical performance and ‘perceived mental’ performance

Appetite suppressant from 5-HT effect

Overdose

  • Stimulant effects: tremors, confusion, dizziness, time passes quickly
  • Cardiac effects: hyperthermia, tachycardia, increased blood pressure, vascular collapse - death
  • Psychotic effects: amphetamine psychosis - hallucinations
74
Q

Discuss illicit ecstasy as CNS stimulant and hallucinogenic

A

Ecstasy / MNDA is a stimulant and hallucinogenic that increases the release of dopamine and serotonin (indirectly acting sympathomimetic)

  • are less effective than amphetamines and LSD
  • feelings of love, closeness and empathy

Adverse effects:

  • Psychological dependence
  • Degeneration of 5-HT and DA neurons
    • disrupts thermoregulation
    • affects mood, sleep, memory and appetite
75
Q

Discuss illicit LSD as a Hallucinogen

A

LSD is a 5-HT2 receptor agonist that results in severe hallucinations

Hallucinogenic effects result from activation of 5-HT2autoreceptors on 5-HT neurones in Raphe

The hallucinations involve confusement of sensory modalities leading to visual, auditory & tactile hallucinations

Less dependence than other drugs -> the experience is so disturbing that adversive

LSD displayes tolerance -> need more to achieve same effects in subsequent use

  • also demonstrates cross-tolerance with other psychomimetics -> need more of them while concurrently on LSD
76
Q

Discuss the effects of caffeine as a CNS stimulant

A

Caffeine is an adenosine antagonist and phosphodiesterases inhibitor; resulting in elevated levels of NA, DA and 5-HT

Increases alertness by stimulating mental activity

  • High doses = anxiety, tension & tremors

No strong dependence effect in animals -> the social aspect of coffee drinking is the factor in humans

77
Q

Describe the effects of Ethanol as a CNS depressant

A

Ethanol is CNS depressant that acts on inhibitory neural pathways including:

  • Inhibit Ca2+channel opening
  • Enhance GABA / GABA-A receptor activity
  • Inhibt glutamate NMDA channels

Effects:

  • Increased self confidence, euphoria, aggression
  • Loss of motor coordination and speech
  • Liver damages, foetal impairment etc

Ethanol use -> marked tolerance (switches on liver enzymes)

Physical dependance

78
Q

Discuss **delta-9 THC **as a CNS depressant

A

**delta-9 THC **is a CNS depressant that acts at cannabinoid receptors of the CNS

Cannabinoid receptor activation results in inhibition of adenylate cyclase -> results in inhibition of neural transmission

CB1-central

  • Impaired short term memory
  • Impaired motor coordination
  • Catalepsy
  • Analgesia
  • Anti-emetic
  • Increased appetite

CB2-peripheral

  • Tachycardia, sympathetic
  • Vasodilatation
  • Reduced intraocular pressure
  • Bronchodilatation
79
Q

Describe the generational development of anti-depressant drugs (for unipolar depression)

A

1st Generation

  • Tricyclic antidepressants
  • MAO inhibitors

2nd Generation

  • Selective Serotonin uptake inhibitors (SSRIs)
  • Selective Serotonin/Noradrenaline uptake inhibitors (SSNRIs)

3rd Generation

  • Novel monoaminergic drugs
  • Non-monoaminergic drugs
80
Q

Discuss tricyclic antidepressants as a treatment for depression

A

TCAs inhibit neuronal uptake of noradrenaline and serotonin

This is achieved by competitively antagonising a-adrenoreceptors and serotonin receptors involved in neural re-uptake.

The drugs have poor selectivity and can affect other receptors including muscarininc and histamine receptors

**Clinical effects took weeks to develop =slow; ** despite pharmacologically effects manifesting in hours -> patients lost complience

Has a narrow therapeutic window

Side effects:

  • Autonomic / CV side effects
  • Weight gain
  • Sedation
  • Seizures
81
Q

Discuss MAO inhibitors as an antidepressant

A

MAO inhibitors inhibit the MAO enzymes responsible for degrading 5-HT, NA and DA

This increases levels of these monamines

This can be either irreversible or reversible

Irreversible

  • ‘Cheese reaction’
    • foods containing the amine tyramine cause a hypertensive crisis due poor metabolism of it

Reversible

  • Less likely to cause cheese reaction
  • Side effects: postural hypotension, dizziness, nausea
82
Q

Discuss selective serotonin reuptake inhibitors as a treatment of depression (SSRIs)

A

Demonstrate selectivity with respect to 5-HT over noradrenaline inhibiting re-uptake.

Large therapeutic window

They are less likely than TCAs to cause anticholinergic side effects and are less dangerous in overdose.

In contrast to MAOIs, they do not cause ‘cheese reactions’.

Side effects:

  • Nausea, insomnia, agitation, weight change, loss of libido
83
Q

Illustrate the progression in anti-depressant development throughout recent history

A
84
Q
A